Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
FASEB J ; 38(2): e23373, 2024 02.
Article in English | MEDLINE | ID: mdl-38217376

ABSTRACT

Fatigue is a common phenomenon closely related to physical discomfort and numerous diseases, which is severely threatening the life quality and health of people. However, the exact mechanisms underlying fatigue are not fully characterized. Herein, we demonstrate that oxaloacetic acid (OAA), a crucial tricarboxylic acid cycle intermediate, modulates the muscle fatigue. The results showed that serum OAA level was positively correlated with fatigue state of mice. OAA-treated induced muscle fatigue impaired the exercise performance of mice. Mechanistically, OAA increased the c-Jun N-terminal kinase (JNK) phosphorylation and uncoupling protein 2 (UCP2) levels in skeletal muscle, which led to decreased energy substrate and enhanced glycolysis. On the other hand, OAA boosted muscle mitochondrial oxidative phosphorylation uncoupled with energy production. In addition, either UCP2 knockout or JNK inhibition totally reversed the effects of OAA on skeletal muscle. Therein, JNK mediated UCP2 activation with OAA-treated. Our studies reveal a novel role of OAA in skeletal muscle metabolism, which would shed light on the mechanism of muscle fatigue and weakness.


Subject(s)
Mitochondria , Oxaloacetic Acid , Humans , Mice , Animals , Oxaloacetic Acid/metabolism , Oxaloacetic Acid/pharmacology , Mitochondria/metabolism , Oxidative Phosphorylation , Citric Acid Cycle , Muscle, Skeletal/metabolism , Uncoupling Protein 2/genetics , Uncoupling Protein 2/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Uncoupling Protein 3/metabolism , Energy Metabolism
2.
Am J Physiol Cell Physiol ; 324(6): C1236-C1248, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37125774

ABSTRACT

We previously found that skeletal muscle mitochondria incubated at low membrane potential (ΔΨ) or interscapular brown adipose tissue (IBAT) mitochondria, wherein ΔΨ is intrinsically low, accumulate oxaloacetate (OAA) in amounts sufficient to inhibit complex II respiration. We proposed a mechanism wherein low ΔΨ reduces reverse electron transport (RET) to complex I causing a low NADH/NAD+ ratio favoring malate conversion to OAA. To further assess the mechanism and its physiologic relevance, we carried out studies of mice with inherently different levels of IBAT mitochondrial inner membrane potential. Isolated complex II (succinate)-energized IBAT mitochondria from obesity-resistant 129SVE mice compared with obesity-prone C57BL/6J displayed greater UCP1 expression, similar O2 flux despite lower ΔΨ, similar OAA concentrations, and similar NADH/NAD+. When GDP was added to inhibit UCP1, 129SVE IBAT mitochondria, despite their lower ΔΨ, exhibited much lower respiration, twofold greater OAA concentrations, much lower RET (as marked by ROS), and much lower NADH and NADH/NAD+ ratios compared with the C57BL/6J IBAT mitochondria. UCP1 knock-out abolished OAA accumulation by succinate-energized mitochondria associated with markedly greater ΔΨ, ROS, and NADH, but equal or greater O2 flux compared with WT mitochondria. GDP addition, compared with no GDP, increased ΔΨ and complex II respiration in wild-type (WT) mice associated with much less OAA. Respiration on complex I substrates followed the more classical dynamics of greater respiration at lower ΔΨ. These findings support the abovementioned mechanism for OAA- and ΔΨ-dependent complex II respiration and support its physiological relevance.NEW & NOTEWORTHY We examined mitochondrial respiration initiated at mitochondrial complex II in mice with varying degrees of brown adipose tissue UCP1 expression. We show that, by affecting inner membrane potential, UCP1 expression determines reverse electron transport from complex II to complex I and, consequently, the NADH/NAD+ ratio. Accordingly, this regulates the level of oxaloacetate accumulation and the extent of oxaloacetate inhibition of complex II.


Subject(s)
Adipose Tissue, Brown , NAD , Mice , Animals , Adipose Tissue, Brown/metabolism , NAD/metabolism , Oxaloacetic Acid/metabolism , Oxaloacetic Acid/pharmacology , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL , Respiration , Obesity/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Membrane Potential, Mitochondrial , Succinates/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
3.
Sheng Li Xue Bao ; 74(4): 505-512, 2022 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-35993201

ABSTRACT

The purpose of the present study was to investigate the effect of glutamate scavenger oxaloacetate (OA) combined with CGS21680, an adenosine A2A receptor (A2AR) agonist, on acute traumatic brain injury (TBI), and to elucidate the underlying mechanisms. C57BL/6J mice were subjected to moderate-level TBI by controlled cortical impact, and then were treated with OA, CGS21680, or OA combined with CGS21680 at acute stage of TBI. At 24 h post TBI, neurological severity score, brain water content, glutamate concentration in cerebrospinal fluid (CSF), mRNA and protein levels of IL-1ß and TNF-α, mRNA level and activity of glutamate oxaloacetate aminotransferase (GOT), and ATP level of brain tissue were detected. The results showed that neurological deficit, brain water content, glutamate concentration in CSF, and the inflammatory cytokine IL-1ß and TNF-α production were exacerbated in CGS21680 treated mice. Administrating OA suppressed the rise of both glutamate concentration in CSF and brain water content, and elevated the ATP level of cerebral tissue. More interestingly, neurological deficit, brain edema, glutamate concentration, IL-1ß and TNF-α levels were ameliorated significantly in mice treated with OA combined with CGS21680. The combined treatment exhibited better therapeutic effects than single OA treatment. We also observed that GOT activity was enhanced in single CGS21680 treatment group, and both the GOT mRNA level and GOT activity were up-regulated in early-stage combined treatment group. These results suggest that A2AR can improve the efficiency of GOT and potentiate the ability of OA to metabolize glutamate. This may be the mechanism that A2AR activation in combination group augmented the neuroprotective effect of OA rather than aggravated the brain damages. Taken together, the present study provides a new insight for the clinical treatment of TBI with A2AR agonists and OA.


Subject(s)
Adenosine A2 Receptor Agonists , Brain Injuries, Traumatic , Neuroprotective Agents , Oxaloacetic Acid , Receptor, Adenosine A2A , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Agonists/therapeutic use , Adenosine Triphosphate , Animals , Brain Injuries/drug therapy , Brain Injuries/genetics , Brain Injuries/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Glutamic Acid , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxaloacetic Acid/pharmacology , Oxaloacetic Acid/therapeutic use , RNA, Messenger , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Tumor Necrosis Factor-alpha/genetics , Water
4.
Sci Rep ; 11(1): 14644, 2021 07 19.
Article in English | MEDLINE | ID: mdl-34282238

ABSTRACT

Inhibition of extracellular glutamate (Glu) release decreases proliferation and invasion, induces apoptosis, and inhibits melanoma metastatic abilities. Previous studies have shown that Blood-glutamate scavenging (BGS), a novel treatment approach, has been found to be beneficial in attenuating glioblastoma progression by reducing brain Glu levels. Therefore, in this study we evaluated the ability of BGS treatment to inhibit brain metastatic melanoma progression in-vivo. RET melanoma cells were implanted in C56BL/6J mice to induce brain melanoma tumors followed by treatment with BGS or vehicle administered for fourteen days. Bioluminescent imaging was conducted to evaluate tumor growth, and plasma/CSF Glu levels were monitored throughout. Immunofluorescence staining of Ki67 and 53BP1 was used to analyze tumor cell proliferation and DNA double-strand breaks. In addition, we analyzed CD8, CD68, CD206, p-STAT1 and iNOS expression to evaluate alterations in tumor micro-environment and anti-tumor immune response due to treatment. Our results show that BGS treatment reduces CSF Glu concentration and consequently melanoma growth in-vivo by decreasing tumor cell proliferation and increasing pro-apoptotic signaling in C56BL/6J mice. Furthermore, BGS treatment supported CD8+ cell recruitment and CD68+ macrophage invasion. These findings suggest that BGS can be of potential therapeutic relevance in the treatment of metastatic melanoma.


Subject(s)
Aspartate Aminotransferase, Cytoplasmic/administration & dosage , Brain Neoplasms/drug therapy , Glutamic Acid/metabolism , Melanoma/drug therapy , Oxaloacetic Acid/administration & dosage , Animals , Apoptosis/drug effects , Aspartate Aminotransferase, Cytoplasmic/pharmacology , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Therapy, Combination , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/secondary , Humans , Melanoma/pathology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Oxaloacetic Acid/pharmacology , Recombinant Proteins/administration & dosage , Signal Transduction/drug effects , Tumor Microenvironment/drug effects
5.
Sci Rep ; 11(1): 11051, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34040085

ABSTRACT

Amyotrophic lateral sclerosis (ALS) remains a devastating motor neuron disease with limited treatment options. Oxaloacetate treatment has a neuroprotective effect in rodent models of seizure and neurodegeneration. Therefore, we treated the ALS model superoxide dismutase 1 (SOD1) G93A mice with oxaloacetate and evaluated their neuromuscular function and lifespan. Treatment with oxaloacetate beginning in the presymptomatic stage significantly improved neuromuscular strength measured during the symptomatic stage in the injected mice compared to the non-treated group. Oxaloacetate treatment starting in the symptomatic stage significantly delayed limb paralysis compared with the non-treated group. For lifespan analysis, oxaloacetate treatment did not show a statistically significant positive effect, but the treatment did not shorten the lifespan. Mechanistically, SOD1G93A mice showed increased levels of tumor necrosis factor-α (TNFα) and peroxisome proliferative activated receptor gamma coactivator 1α (PGC-1α) mRNAs in the spinal cord. However, oxaloacetate treatment reverted these abnormal levels to that of wild-type mice. Similarly, the altered expression level of total NF-κB protein returned to that of wild-type mice with oxaloacetate treatment. These results suggest that the beneficial effects of oxaloacetate treatment in SOD1G93A mice may reflect the effects on neuroinflammation or bioenergetic stress.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Motor Activity/drug effects , Oxaloacetic Acid/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Spinal Cord/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Disease Models, Animal , Inflammation/drug therapy , Inflammation/metabolism , Longevity/drug effects , Mice , Motor Neurons/drug effects , Motor Neurons/metabolism , Oxaloacetic Acid/therapeutic use , Spinal Cord/metabolism , Superoxide Dismutase/metabolism
6.
Biochem Biophys Res Commun ; 528(2): 318-321, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32475641

ABSTRACT

Staphylococcus aureus is a resident skin bacterium involved in the exacerbation of atopic dermatitis. Here we report that S. aureus regulates the tricarboxylic acid (TCA) cycle via the production of pyruvate for tolerance to betamethasone valerate (BV), an anti-inflammatory drug used in the treatment of atopic dermatitis. The addition of BV or clobetasol propionate to the medium among 5 different anti-inflammatory steroids delayed the growth of S. aureus. Comprehensive gene expression analysis by RNA-seq revealed that BV increased the expression of genes related to glycolysis in S. aureus. Pyruvate, a product of glycolysis, suppressed the S. aureus growth inhibition by BV. The addition of oxaloacetate, a compound in the TCA cycle biosynthesized from pyruvate, was also suppressed the inhibitory effect of BV. Malonate, an inhibitor of succinate dehydrogenase in the TCA cycle, increased the inhibitory effect of BV on the growth of S. aureus. These findings suggest that S. aureus promotes tolerance to BV, an anti-inflammatory steroid, by regulating the TCA cycle via the production of pyruvate.


Subject(s)
Betamethasone Valerate/toxicity , Citric Acid Cycle/drug effects , Pyruvic Acid/pharmacology , Staphylococcus aureus/metabolism , Malonates/pharmacology , Oxaloacetic Acid/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Steroids/pharmacology
7.
Mini Rev Med Chem ; 20(10): 818-830, 2020.
Article in English | MEDLINE | ID: mdl-31902356

ABSTRACT

Pyrrole is a very important pharmacophoric moiety. It has been widely incorporated into the skeleton of antitumor, anti-inflammatory, antibacterial, antioxidant and antifungal active substances. Access to this key heterocycle by diverse routes is particularly attractive in terms of chemistry, and also from the environmental point of view. The present minireview summarizes the reported methods for the preparation of highly substituted pyrrole derivatives based on the one-pot multicomponent reaction of aldehydes, primary amines, and oxalacetate analogues as well as their biology.


Subject(s)
Aldehydes/chemistry , Aldehydes/pharmacology , Amines/chemistry , Amines/pharmacology , Oxaloacetic Acid/chemistry , Oxaloacetic Acid/pharmacology , Pyrroles/chemistry , Drug Discovery
8.
J Biol Chem ; 293(51): 19932-19941, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30385511

ABSTRACT

We recently reported a previously unrecognized mitochondrial respiratory phenomenon. When [ADP] was held constant ("clamped") at sequentially increasing concentrations in succinate-energized muscle mitochondria in the absence of rotenone (commonly used to block complex I), we observed a biphasic, increasing then decreasing, respiratory response. Here we investigated the mechanism. We confirmed decades-old reports that oxaloacetate (OAA) inhibits succinate dehydrogenase (SDH). We then used an NMR method to assess OAA concentrations (known as difficult to measure by MS) as well as those of malate, fumarate, and citrate in isolated succinate-respiring mitochondria. When these mitochondria were incubated at varying clamped ADP concentrations, respiration increased at low [ADP] as expected given the concurrent reduction in membrane potential. With further increments in [ADP], respiration decreased associated with accumulation of OAA. Moreover, a low pyruvate concentration, that alone was not enough to drive respiration, was sufficient to metabolize OAA to citrate and completely reverse the loss of succinate-supported respiration at high [ADP]. Further, chemical or genetic inhibition of pyruvate uptake prevented OAA clearance and preserved respiration. In addition, we measured the effects of incremental [ADP] on NADH, superoxide, and H2O2 (a marker of reverse electron transport from complex II to I). In summary, our findings, taken together, support a mechanism (detailed within) wherein succinate-energized respiration as a function of increasing [ADP] is initially increased by [ADP]-dependent effects on membrane potential but subsequently decreased at higher [ADP] by inhibition of succinate dehydrogenase by OAA. The physiologic relevance is discussed.


Subject(s)
Adenosine Diphosphate/metabolism , Electron Transport Complex II/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Oxaloacetic Acid/pharmacology , Animals , Cell Respiration/drug effects , Electron Transport Complex II/metabolism , Energy Metabolism/drug effects , Mitochondria/enzymology , Muscle Cells/cytology , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
9.
Cell Death Dis ; 9(11): 1135, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30429455

ABSTRACT

Cell models of mitochondrial complex I (CI) deficiency display activation of glycolysis to compensate for the loss in mitochondrial ATP production. This adaptation can mask other relevant deficiency-induced aberrations in cell physiology. Here we investigated the viability, mitochondrial morphofunction, ROS levels and ATP homeostasis of primary skin fibroblasts from Leigh Syndrome (LS) patients with isolated CI deficiency. These cell lines harbored mutations in nuclear DNA (nDNA)-encoded CI genes (NDUFS7, NDUFS8, NDUFV1) and, to prevent glycolysis upregulation, were cultured in a pyruvate-free medium in which glucose was replaced by galactose. Following optimization of the cell culture protocol, LS fibroblasts died in the galactose medium, whereas control cells did not. LS cell death was dose-dependently inhibited by pyruvate, malate, oxaloacetate, α-ketoglutarate, aspartate, and exogenous NAD+ (eNAD), but not by lactate, succinate, α-ketobutyrate, and uridine. Pyruvate and eNAD increased the cellular NAD+ content in galactose-treated LS cells to a different extent and co-incubation studies revealed that pyruvate-induced rescue was not primarily mediated by NAD+. Functionally, in LS cells glucose-by-galactose replacement increased mitochondrial fragmentation and mass, depolarized the mitochondrial membrane potential (Δψ), increased H2DCFDA-oxidizing ROS levels, increased mitochondrial ATP generation, and reduced the total cellular ATP content. These aberrations were differentially rescued by pyruvate and eNAD, supporting the conclusion that these compounds rescue galactose-induced LS cell death via different mechanisms. These findings establish a cell-based strategy for intervention testing and enhance our understanding of CI deficiency pathophysiology.


Subject(s)
Electron Transport Complex I/deficiency , Fibroblasts/drug effects , Galactose/antagonists & inhibitors , Leigh Disease/metabolism , Mitochondrial Diseases/genetics , NAD/pharmacology , Pyruvic Acid/pharmacology , Adenosine Triphosphate/biosynthesis , Aspartic Acid/metabolism , Aspartic Acid/pharmacology , Cell Death/drug effects , Culture Media/chemistry , Culture Media/pharmacology , Electron Transport Complex I/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Galactose/metabolism , Galactose/pharmacology , Gene Expression , Glycolysis/drug effects , Humans , Ketoglutaric Acids/metabolism , Ketoglutaric Acids/pharmacology , Leigh Disease/genetics , Leigh Disease/pathology , Malates/metabolism , Malates/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mutation , NAD/metabolism , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Oxaloacetic Acid/metabolism , Oxaloacetic Acid/pharmacology , Primary Cell Culture , Pyruvic Acid/metabolism , Skin/drug effects , Skin/metabolism , Skin/pathology
10.
PLoS Pathog ; 14(10): e1007295, 2018 10.
Article in English | MEDLINE | ID: mdl-30308066

ABSTRACT

Amebiasis, a global intestinal parasitic disease, is due to Entamoeba histolytica. This parasite, which feeds on bacteria in the large intestine of its human host, can trigger a strong inflammatory response upon invasion of the colonic mucosa. Whereas information about the mechanisms which are used by the parasite to cope with oxidative and nitrosative stresses during infection is available, knowledge about the contribution of bacteria to these mechanisms is lacking. In a recent study, we demonstrated that enteropathogenic Escherichia coli O55 protects E. histolytica against oxidative stress. Resin-assisted capture (RAC) of oxidized (OX) proteins coupled to mass spectrometry (OX-RAC) was used to investigate the oxidation status of cysteine residues in proteins present in E. histolytica trophozoites incubated with live or heat-killed E. coli O55 and then exposed to H2O2-mediated oxidative stress. We found that the redox proteome of E. histolytica exposed to heat-killed E. coli O55 is enriched with proteins involved in redox homeostasis, lipid metabolism, small molecule metabolism, carbohydrate derivative metabolism, and organonitrogen compound biosynthesis. In contrast, we found that proteins associated with redox homeostasis were the only OX-proteins that were enriched in E. histolytica trophozoites which were incubated with live E. coli O55. These data indicate that E. coli has a profound impact on the redox proteome of E. histolytica. Unexpectedly, some E. coli proteins were also co-identified with E. histolytica proteins by OX-RAC. We demonstrated that one of these proteins, E. coli malate dehydrogenase (EcMDH) and its product, oxaloacetate, are key elements of E. coli-mediated resistance of E. histolytica to oxidative stress and that oxaloacetate helps the parasite survive in the large intestine. We also provide evidence that the protective effect of oxaloacetate against oxidative stress extends to Caenorhabditis elegans.


Subject(s)
Entamoeba histolytica/drug effects , Entamoebiasis/drug therapy , Escherichia coli/physiology , Oxaloacetic Acid/pharmacology , Oxidative Stress/drug effects , Protozoan Proteins/metabolism , Amebiasis/drug therapy , Amebiasis/metabolism , Amebiasis/parasitology , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/parasitology , Cells, Cultured , Entamoebiasis/metabolism , Entamoebiasis/parasitology , HeLa Cells , Humans , Intestine, Large/drug effects , Intestine, Large/metabolism , Intestine, Large/parasitology , Macrophages/cytology , Macrophages/drug effects , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA
11.
Int J Mol Sci ; 19(6)2018 May 31.
Article in English | MEDLINE | ID: mdl-29857490

ABSTRACT

Chemical injury is partly due to free radical lipid peroxidation, which can induce oxidative stress and produce a large number of reactive oxygen species (ROS). Oxaloacetic acid is an important intermediary in the tricarboxylic acid cycle (TCA cycle) and participates in metabolism and energy production. In our study, we found that oxaloacetate (OA) effectively alleviated liver injury which was induced by hydrogen peroxide (H2O2) in vitro and carbon tetrachloride (CCl4) in vivo. OA scavenged ROS, prevented oxidative damage and maintained the normal structure of mitochondria. We further confirmed that OA increased adenosine triphosphate (ATP) by promoting the TCA production cycle and oxidative phosphorylation (OXPHOS). Finally, OA inhibited the mitogen-activated protein kinase (MAPK) and apoptotic pathways by suppressing tumor necrosis factor-α (TNF-α). Our findings reveal a mechanism for OA ameliorating chemical liver injury and suggest a possible implementation for preventing the chemical liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Energy Metabolism/drug effects , Oxaloacetic Acid/pharmacology , Oxidative Stress/drug effects , Animals , Antioxidants/pharmacology , Carbon Tetrachloride/adverse effects , Chemical and Drug Induced Liver Injury/drug therapy , Citric Acid Cycle/drug effects , Disease Models, Animal , Glycolysis , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hydrogen Peroxide/metabolism , Lipid Peroxidation/drug effects , Mice , Mitochondria/metabolism , Models, Biological , Oxidative Phosphorylation/drug effects , Protective Agents/pharmacology
12.
Cancer Med ; 7(4): 1416-1429, 2018 04.
Article in English | MEDLINE | ID: mdl-29533007

ABSTRACT

Most cancer cells perform glycolysis despite having sufficient oxygen. The specific metabolic pathways of cancer cells have become the focus of cancer treatment. Recently, accumulating evidence indicates oxidative phosphorylation (OXPHOS) and glycolysis can be regulated with each other. Thus, we suggest that the glycolysis of cancer cells is inhibited by restoring or improving OXPHOS in cancer cells. In our study, we found that oxaloacetate (OA) induced apoptosis in HepG2 cells in vivo and in vitro. Meanwhile, we found that OA induced a decrease in the energy metabolism of HepG2 cells. Further results showed that the expression and activity of glycolytic enzymes were decreased with OA treatment. Conversely, the expression and activity of enzymes involved in the TCA cycle and OXPHOS were increased with OA treatment. The results indicate that OA can inhibit glycolysis through enhancement of OXPHOS. In addition, OA-mediated suppression of HIF1α, p-Akt, and c-myc led to a decrease in glycolysis level. Therefore, OA has the potential to be a novel anticancer drug.


Subject(s)
Apoptosis/drug effects , Oxaloacetic Acid/pharmacology , Animals , Citric Acid Cycle/drug effects , Energy Metabolism/drug effects , Female , Glycolysis/drug effects , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Models, Biological , Mutation , Oxidative Phosphorylation/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
13.
J Inorg Biochem ; 178: 63-69, 2018 01.
Article in English | MEDLINE | ID: mdl-29078149

ABSTRACT

Non-heme Fe(II)/α-ketoglutarate (αKG)-dependent oxygenases catalyze a wide array of reactions through coupling oxidative decarboxylation of αKG to substrate oxygenation. This class of enzymes follows a sequential mechanism in which O2 reacts only after binding primary substrate, raising questions over how protein structure tailors molecular access to the Fe(II) cofactor. The enzyme "factor inhibiting hypoxia inducible factor" (FIH) senses pO2 in human cells by hydroxylating the C-terminal transactivation domain (CTAD), suggesting that structural elements limiting molecular access to the active site may limit the pO2 response. In this study, we tested the impact of a solvent-accessible tunnel in FIH on molecular access to the active site in FIH. The size of the tunnel was increased through alanine point mutagenesis (Y93A, E105A, and Q147A), followed by a suite of mechanistic and spectroscopic probes. Steady-state kinetics varying O2 or CTAD indicated that O2 passage through the tunnel was not affected by Ala substitutions, allowing us to conclude that this narrow tunnel did not impact pO2 sensing by FIH. Steady-state kinetics with varied αKG concentrations revealed increased substrate inhibition for the Ala variants, suggesting that a second αKG molecule may bind near the active site of FIH. If this solvent-accessible tunnel is the O2 entry tunnel, it may be narrow in order to permit O2 access while preventing metabolic intermediates, such as αKG, from inhibiting FIH under physiological conditions.


Subject(s)
Hypoxia-Inducible Factor 1/antagonists & inhibitors , Hypoxia-Inducible Factor 1/chemistry , Ketoglutaric Acids/metabolism , Oxygenases/metabolism , Catalytic Domain , Citric Acid/chemistry , Citric Acid/pharmacology , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Genetic Variation , Humans , Hypoxia-Inducible Factor 1/genetics , Ketoglutaric Acids/chemistry , Kinetics , Oxaloacetic Acid/chemistry , Oxaloacetic Acid/pharmacology , Oxygenases/chemistry , Solvents/chemistry
14.
Int J Biol Macromol ; 101: 59-66, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28322963

ABSTRACT

Oxaloacetic acid (OA) is naturally found in organisms and well known as an intermediate of citric acid cycle producing ATP. We evaluated the effects of OA on tyrosinase activity and structure via integrating methods of enzyme kinetics and computational simulations. OA was found to be a reversible inhibitor of tyrosinase and its induced mechanism was the parabolic non-competitive inhibition type (IC50=17.5±0.5mM and Ki=6.03±1.36mM). Kinetic measurements by real-time interval assay showed that OA induced multi-phasic inactivation process composing with fast (k1) and slow (k2) phases. Spectrofluorimetry studies showed that OA mainly induced regional changes in the active site of tyrosinase accompanying with hydrophobic disruption at high dose. The computational docking simulations further revealed that OA could interact with several residues near the tyrosinase active site pocket such as HIS61, HIS259, HIS263, and VAL283. Our study provides insight into the mechanism by which energy producing intermediate such as OA inhibit tyrosinase and OA is a potential natural anti-pigmentation agent.


Subject(s)
Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/chemistry , Oxaloacetic Acid/metabolism , Oxaloacetic Acid/pharmacology , Agaricus/enzymology , Catalytic Domain/drug effects , Kinetics , Monophenol Monooxygenase/metabolism , Safety
15.
Neoplasma ; 64(2): 192-198, 2017.
Article in English | MEDLINE | ID: mdl-28043145

ABSTRACT

Oxaloacetate (OA) is one of the intermediates of the Krebs cycle. In addition to its role in energy production, OA may have other effects on the cell. We report here that OA could have a cell type dependent cytotoxic effect on the human hepatic carcinoma cell line HepG2 through induction of apoptosis and reactive oxygen species (ROS) accumulation. In our study, OA decreased the viability and colony formation of HepG2 cells and induced cell death. Caspase-3 activity was increased, the pro-apoptotic protein Bax was up-regulated, and the anti-apoptotic protein Bcl-2 was down-regulated in OA-treated HepG2 cells indicating that apoptosis through the intrinsic pathway was involved in the cell death. The ROS level in OA-treated HepG2 cells was increased. The anti-oxidant N-acetylcysteine (NAC) and glutathione (GSH) prevented the OA-induced decrease in cell but did not alter the enhanced apoptotic Bax/Bcl-2 mRNA ratio. These results suggest that the OA-induced apoptosis of HepG2 cell is not driven by oxidative damage and at least two distinct mechanisms, one mediated by ROS and one involving apoptosis, result in the cytotoxic effects of OA on HepG2 cells. These studies expand the biological functional repertoire of OA and provide a mechanism by which hepatocellular carcinoma may be targeted by OA.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Oxaloacetic Acid/pharmacology , Reactive Oxygen Species/metabolism , Acetylcysteine , Antineoplastic Agents/pharmacology , Caspase 3/metabolism , Glutathione , Hep G2 Cells/drug effects , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism
16.
Cereb Cortex ; 27(7): 3724-3735, 2017 07 01.
Article in English | MEDLINE | ID: mdl-27390019

ABSTRACT

Dysregulation of glutamate homeostasis in the interstitial fluid of the brain is strongly implicated in causing synaptic dysfunction in many neurological and psychiatric illnesses. In the case of Alzheimer's disease (AD), amyloid ß (Aß)-mediated disruption of synaptic plasticity and memory can be alleviated by interventions that directly remove glutamate or block certain glutamate receptors. An alternative strategy is to facilitate the removal of excess glutamate from the nervous system by activating peripheral glutamate clearance systems. One such blood-based system, glutamate oxaloacetate transaminase (GOT), is activated by oxaloacetate, which acts as a co-substrate. We report here that synthetic and AD brain-derived Aß-mediated inhibition of synaptic long-term potentiation in the hippocampus is alleviated by oxaloacetate. Moreover the effect of oxaloacetate was GOT-dependent. The disruptive effects of a general inhibitor of excitatory amino acid transport or TNFα, a pro-inflammatory mediator of Aß action, were also reversed by oxaloacetate. Furthermore, another intervention that increases peripheral glutamate clearance, peritoneal dialysis, mimicked the beneficial effect of oxaloacetate. These findings lend support to the promotion of the peripheral clearance of glutamate as a means to alleviate synaptic dysfunction that is caused by impaired glutamate homeostasis in the brain.


Subject(s)
Amyloid beta-Peptides/pharmacology , Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/blood , Hippocampus/metabolism , Homeostasis/physiology , Synapses/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Aspartate Aminotransferase, Cytoplasmic/pharmacology , Aspartic Acid/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Female , Hippocampus/drug effects , Homeostasis/drug effects , Humans , Injections, Intraperitoneal , Male , Oxaloacetic Acid/pharmacology , Peptide Fragments/pharmacology , Rats , Rats, Wistar , Synapses/physiology , Time Factors , Tumor Necrosis Factor-alpha/metabolism
17.
Brain Res Bull ; 128: 7-12, 2017 01.
Article in English | MEDLINE | ID: mdl-27816553

ABSTRACT

Adipose stromal cells conditioned media (ASC-CM) protect neurons in a variety of neuronal death models including potassium/serum deprivation-induced neuronal apoptosis. In this study, we found that ASC-CM contained glutamate oxaloacetate transaminase and its substrate, oxaloacetate (OAA) directly protected cerebellar granule neurons (CGN) from apoptosis induced by serum and potassium deprivation. Additionally, OAA inhibited serum and potassium deprivation-induced caspase 3 activation. ASC-CM and OAA in combination had a synergistic neuroprotective effect. Clearly, different from ASC-CM-induced neuroprotection, OAA-induced neuroprotection was Akt- independent but JNK-dependent. These data establish a mechanistic basis supporting that the application of ASC-CM for neuroprotective treatments could be significantly enhanced by addition of OAA.


Subject(s)
Adipose Tissue/cytology , Apoptosis , Neurons/physiology , Neuroprotective Agents/pharmacology , Oxaloacetic Acid/pharmacology , Stromal Cells/physiology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cells, Cultured , Cerebellum/cytology , Culture Media, Conditioned , Culture Media, Serum-Free , Humans , MAP Kinase Kinase 4/metabolism , Neurons/drug effects , Potassium/analysis , Rats, Sprague-Dawley , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism
18.
Biochim Biophys Acta ; 1857(9): 1561-1568, 2016 09.
Article in English | MEDLINE | ID: mdl-27287543

ABSTRACT

Mitochondrial Complex II is a key mitochondrial enzyme connecting the tricarboxylic acid (TCA) cycle and the electron transport chain. Studies of complex II are clinically important since new roles for this enzyme have recently emerged in cell signalling, cancer biology, immune response and neurodegeneration. Oxaloacetate (OAA) is an intermediate of the TCA cycle and at the same time is an inhibitor of complex II with high affinity (Kd~10(-8)M). Whether or not OAA inhibition of complex II is a physiologically relevant process is a significant, but still controversial topic. We found that complex II from mouse heart and brain tissue has similar affinity to OAA and that only a fraction of the enzyme in isolated mitochondrial membranes (30.2±6.0% and 56.4±5.6% in the heart and brain, respectively) is in the free, active form. Since OAA could bind to complex II during isolation, we established a novel approach to deplete OAA in the homogenates at the early stages of isolation. In heart, this treatment significantly increased the fraction of free enzyme, indicating that OAA binds to complex II during isolation. In brain the OAA-depleting system did not significantly change the amount of free enzyme, indicating that a large fraction of complex II is already in the OAA-bound inactive form. Furthermore, short-term ischemia resulted in a dramatic decline of OAA in tissues, but it did not change the amount of free complex II. Our data show that in brain OAA is an endogenous effector of complex II, potentially capable of modulating the activity of the enzyme.


Subject(s)
Brain/enzymology , Electron Transport Complex II/antagonists & inhibitors , Mitochondria/enzymology , Myocardium/enzymology , Oxaloacetic Acid/pharmacology , Animals , Mice , Succinate Dehydrogenase/antagonists & inhibitors , Succinate Dehydrogenase/metabolism
19.
J Neurochem ; 137(1): 76-87, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26811028

ABSTRACT

We tested how the addition of oxaloacetate (OAA) to SH-SY5Y cells affected bioenergetic fluxes and infrastructure, and compared the effects of OAA to malate, pyruvate, and glucose deprivation. OAA displayed pro-glycolysis and pro-respiration effects. OAA pro-glycolysis effects were not a consequence of decarboxylation to pyruvate because unlike OAA, pyruvate lowered the glycolysis flux. Malate did not alter glycolysis flux and reduced mitochondrial respiration. Glucose deprivation essentially eliminated glycolysis and increased mitochondrial respiration. OAA increased, while malate decreased, the cell NAD+/NADH ratio. Cytosolic malate dehydrogenase 1 protein increased with OAA treatment, but not with malate or glucose deprivation. Glucose deprivation increased protein levels of ATP citrate lyase, an enzyme which produces cytosolic OAA, whereas OAA altered neither ATP citrate lyase mRNA nor protein levels. OAA, but not glucose deprivation, increased cytochrome oxidase subunit 2, PGC1α, PGC1ß, and PGC1 related co-activator protein levels. OAA increased total and phosphorylated SIRT1 protein. We conclude that adding OAA to SH-SY5Y cells can support or enhance both glycolysis and respiration fluxes. These effects appear to depend, at least partly, on OAA causing a shift in the cell redox balance to a more oxidized state, that it is not a glycolysis pathway intermediate, and possibly its ability to act in an anaplerotic fashion. We examined how oxaloacetate (OAA) affects bioenergetic fluxes. To advance the understanding of how OAA mediates these changes, we compared the effects of OAA to malate, pyruvate, and glucose deprivation. We further examined how OAA affects levels of enzymes that facilitate its cytosolic metabolism, and found OAA increased the expression of malate dehydrogenase 1 (MDH1-cytosolic). We propose the following: OAA supports both glycolysis and respiration fluxes, shifts the cell redox balance toward a more oxidized state, and acts in an anaplerotic fashion. Abbreviations not defined in the text: MDH2, malate dehydrogenase 2 (mitochondrial).


Subject(s)
Mitochondria/drug effects , Neurons/drug effects , Oxaloacetic Acid/pharmacology , Adenosine Triphosphate/metabolism , Cell Line , Cell Line, Tumor , Cytosol/metabolism , Energy Metabolism/drug effects , Glucose/metabolism , Glucose/pharmacology , Glycolysis/drug effects , Humans , Malate Dehydrogenase/metabolism , Malates/metabolism , Malates/pharmacology , Mitochondria/metabolism , NAD/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Oxygen Consumption , Pyruvic Acid/metabolism , Pyruvic Acid/pharmacology , RNA, Messenger/biosynthesis
20.
Curr Top Med Chem ; 16(8): 849-57, 2016.
Article in English | MEDLINE | ID: mdl-26311426

ABSTRACT

Mitochondrial dysfunction and neuroinflammation occur in Alzheimer's disease (AD). The causes of these pathologic lesions remain uncertain, but links between these phenomena are increasingly recognized. In this review, we discuss data that indicate mitochondria or mitochondrial components may contribute to neuroinflammation. While mitochondrial dysfunction could cause neuroinflammation, neuroinflammation could also cause mitochondrial dysfunction. However, based on the systemic nature of AD mitochondrial dysfunction as well as data from experiments we discuss, the former possibility is perhaps more likely. If correct, then manipulation of mitochondria, either directly or through manipulations of bioenergetic pathways, could prove effective in reducing metabolic dysfunction and neuroinflammation in AD patients. We also review some potential approaches through which such manipulations may be achieved.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Inflammation/metabolism , Mitochondria/metabolism , Alzheimer Disease/diet therapy , DNA, Mitochondrial , Energy Metabolism , Exercise , Humans , Inflammation/diet therapy , Inflammation/physiopathology , Ketone Bodies/metabolism , Mitochondria/genetics , Nervous System Diseases/metabolism , Nervous System Diseases/physiopathology , Oxaloacetic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...